WO2018126183A2 - Peptides her3 pour imagerie et radiothérapie - Google Patents

Peptides her3 pour imagerie et radiothérapie Download PDF

Info

Publication number
WO2018126183A2
WO2018126183A2 PCT/US2017/069031 US2017069031W WO2018126183A2 WO 2018126183 A2 WO2018126183 A2 WO 2018126183A2 US 2017069031 W US2017069031 W US 2017069031W WO 2018126183 A2 WO2018126183 A2 WO 2018126183A2
Authority
WO
WIPO (PCT)
Prior art keywords
composition
natural derivative
group
acid
imaging
Prior art date
Application number
PCT/US2017/069031
Other languages
English (en)
Other versions
WO2018126183A3 (fr
Inventor
Umar Mahmood
Benjamin LARIMER
Original Assignee
The General Hospital Corporation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by The General Hospital Corporation filed Critical The General Hospital Corporation
Priority to US16/475,020 priority Critical patent/US11241511B2/en
Publication of WO2018126183A2 publication Critical patent/WO2018126183A2/fr
Publication of WO2018126183A3 publication Critical patent/WO2018126183A3/fr
Priority to US17/562,861 priority patent/US20220175975A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/08Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins
    • A61K51/088Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins conjugates with carriers being peptides, polyamino acids or proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics

Definitions

  • This invention relates to compositions useful for imaging techniques, and more particularly to compounds that are useful for imaging HER3 using medical imaging, including positron emission tomography.
  • the receptor tyrosine kinase HER3 (ERBB3) is upregulated in response to targeted therapies in multiple cancers, including lung, breast and prostate cancers.
  • ERBB3 receptor tyrosine kinase HER3
  • levels of HER3 expression are highly dynamic, making quantification by tissue sampling techniques such as biopsy potentially inaccurate.
  • PET imaging which permits global and serial imaging, may present a more robust method for HER3 quantification, improving HER3 diagnosis, facilitating improved clinical trials assessing HER3 expression, and, in the long term, providing accurate diagnosis of HER3 status to help diagnosis targeted therapy resistance.
  • A comprises an imaging agent
  • B is a linking group or a covalent bond
  • C is a polypeptide comprising from about 9 to about 75 amino acids, wherein the polypeptide comprises an amino acid sequence having at least 90% sequence identity to: Q ID NO: 12) wherein:
  • X 2 is selected from the group consisting of L, L*, I, I*, P, P*, V, V*, G, and G*;
  • X 3 is selected from the group consisting of P, P*, L, L*, I, I*, V, V*, G, and G*;
  • X 4 is selected from the group consisting of T, T*, S, S*, C, C*, M, and M*;
  • X 5 is selected from the group consisting of any L-amino acid, any D-amino acid, and any non-natural amino acid;
  • X 6 is selected from the group consisting of any L-amino acid, any D-amino acid, and any non-natural amino acid;
  • X 7 is selected from the group consisting of R, R*, H, H*, K, K*, N, N*, Q, and Q*, and
  • X 8 is selected from the group consisting of S, S*, T, T*, C, C*, M, and M*, wherein:
  • L* is a non-natural derivative of L
  • I* is a non-natural derivative of I
  • P* is a non-natural derivative of P
  • V* is a non-natural derivative of V
  • G* is a non-natural derivative of G
  • T* is a non-natural derivative of T
  • S* is a non-natural derivative of S
  • C* is a non-natural derivative of C
  • M* is a non-natural derivative of M
  • R* is a non- natural derivative of R
  • H* is a non-natural derivative of H
  • K* is a non-natural derivative of K
  • N* is a non-natural derivative of N
  • Q* is a non-natural derivative of Q.
  • A comprises one or more imaging agents selected from the group consisting of a paramagnetic ion, an x-ray imaging agent, a fluorophore, and a radioisotope.
  • A comprises a radioisotope.
  • the radioisotope is suitable for PET imaging.
  • the radioisotope is selected from the group consisting of 3 ⁇ 4, n C, 14 C, 18 F, 32 P, 35 S, 36 C1, 51 Cr, 52 Fe, 57 Co,
  • the radioisotope selected from the group consisting of 3 H,
  • A comprises b8 Ga.
  • A further comprises a chelating agent.
  • the chelating agent is selected from the group consisting of 1,4,7- triazacyclononanetriacetic acid (NOTA), 1, 4,7,10-tetraazacyclododecane- 1,4,7, 10- tetraacetic acid (DOTA), 1,4,7-triazacyclononane-l-glutaric acid-4,7-diacetic acid (NODAGA), ethylene diamine tetra-acetic acid (EDTA), diethylene triaminepentaacetic acid (DTP A), cyclohexyl-l,2-diaminetetraacetic acid (CDTA), ethyl eneglycol-0,0'- bis(2-aminoethyl)-N,N,N',N'-tetraacetic acid (EGTA), N,N-bis(hydroxybenzyl)- ethylenediamine-N,N'-diacetic acid (HBED), triethylene tetramine hexaacetic acid (TTHA),
  • the chelating agent is selected from the group consisting of 1,4,7- triazacyclononanetriacetic acid (NOTA), 1, 4,7, 10-tetraazacyclododecane- 1,4,7, 10- tetraacetic acid (DOTA), 1,4,7-triazacyclononane-l-glutaric acid-4,7-diacetic acid (NODAGA), and 2,2',2"-(2-(4-isothiocyanatobenzyl)-l,4,7-triazonane-l,4,7-triyl)triacetic acid (NOTA-NCS).
  • NOTA 1,4,7- triazacyclononanetriacetic acid
  • DOTA 1,4,7, 10-tetraazacyclododecane- 1,4,7, 10- tetraacetic acid
  • NODAGA 1,4,7-triazacyclononane-l-glutaric acid-4,7-diacetic acid
  • NCS 2,2',2"-(2-(4-is
  • the chelating agent is 1,4,7- triazacyclononanetriacetic acid (NOTA) or 2,2',2"-(2-(4-isothiocyanatobenzyl)- 1,4,7- triazonane-l,4,7-triyl)triacetic acid (NOTA-NCS).
  • B is a linking group.
  • B is a linking group comprising one or more Ci-30 alkyleneoxy groups, one or more independently selected amino acids, or any combination thereof. In some embodiments, B is a linking group comprising from about 1 to about 10 independently selected amino acids. In some embodiments, B is a linking group comprising a sequence having at least 90% sequence identity to:
  • X 9 is selected from the group consisting of beta A, 6-aminohexnoic acid, 8- aminooctanoic acid, and 2-(2-(2-aminoethoxy)ethoxy)acetic acid;
  • X 10 is selected from the group consisting of G, G*, A, A*, S, S*, P, P*, W, W*, Y, Y*, H, H*, T, T*, M, M*, N, N*, Q, and Q*;
  • X 11 is selected from the group consisting of G, G*, A, A*, S, S*, P, P*, W, W*, Y, Y*, H, H*, T, T*, M, M*, N, N*, Q, and Q*; and
  • X 12 is selected from the group consisting of G, G*, A, A*, S, S*, P, P*, W, W*, Y, Y*, H, H*, T, T*, M, M*, N, N*, Q, and Q*;
  • G* is a non-natural derivative of G
  • A* is a non-natural derivative of A
  • S* is a non-natural derivative of S
  • P* is a non-natural derivative of P
  • W* is a non-natural derivative of W
  • Y* is a non-natural derivative of Y
  • H* is a non-natural derivative of H
  • T* is a non-natural derivative of T
  • M* is a non-natural derivative of M
  • N* is a non-natural derivative of N
  • Q* is a non-natural derivative of Q.
  • B is a linking group which is:
  • « ⁇ / refers to the bond between B and C.
  • C is a polypeptide comprising from about 15 to about 25 amino acids, wherein the polypeptide comprises an amino acid sequence having at least 90% sequence identity to SEQ ID NO: 12.
  • X 2 is L or L*.
  • X 3 is P or P*.
  • X 4 is T or T*.
  • X 5 is K or K* .
  • X 6 is F or F*.
  • X 7 is R or R* .
  • X 8 is S or S*.
  • composition of Formula I is a composition of Formula la:
  • composition of Formula I is a composition of Formula lb:
  • A*-(NOTA-NCS)-beta A-G-G-G-C-X 2 -X 3 -X 4 -X 5 -X 6 -X 7 -X ;
  • A' is a radioisotope
  • the composition of Formula I is a composition of Formula
  • refers to a disulfide bond between the cysteine groups.
  • composition of Formula I is:
  • B comprises from about 1 to about 10 amino acids
  • C is a polypeptide comprising from about 9 to about 75 amino acids, wherein the polypeptide comprises an amino acid sequence having at least 90% sequence identity to: Q ID NO: 12) wherein:
  • ⁇ / refers to the bond between C and B; refers to a disulfide bond between the cysteine groups;
  • X 2 is selected from the group consisting of L, L*, I, I*, P, P*, V, V*, G, and G*;
  • X 3 is selected from the group consisting of P, P*, L, L*, I, I*, V, V*, G, and G*;
  • X 4 is selected from the group consisting of T, T*, S, S*, C, C*, M, and M*;
  • X 5 is selected from the group consisting of any L-amino acid, any D-amino acid, and any non-natural amino acid;
  • X 6 is selected from the group consisting of any L-amino acid, any D-amino acid, and any non-natural amino acid;
  • X 7 is selected from the group consisting of R, R*, H, H*, K, K*, N, N*, Q, and Q*, and
  • X 8 is selected from the group consisting of S, S*, T, T*, C, C*, M, and M*, wherein:
  • L* is a non-natural derivative of L
  • I* is a non-natural derivative of I
  • P* is a non-natural derivative of P
  • V* is a non-natural derivative of V
  • G* is a non-natural derivative of G
  • T* is a non-natural derivative of T
  • S* is a non-natural derivative of S
  • C* is a non-natural derivative of C
  • M* is a non- natural derivative of M
  • R* is a non- natural derivative of R
  • H* is a non-natural derivative of H
  • K* is a non-natural derivative of K
  • N* is a non-natural derivative of N
  • Q* is a non-natural derivative of Q.
  • B comprises a sequence having at least 90% sequence identity to:
  • ⁇ ww refers to the bond between B and C
  • X 9 is selected from the group consisting of beta A, 6-aminohexnoic acid, 8- aminooctanoic acid, and 2-(2-(2-aminoethoxy)ethoxy)acetic acid;
  • X 10 is selected from the group consisting of G, G*, A, A*, S, S*, P, P*, W, W*, Y, Y*, H, H*, T, T*, M, M*, N, N*, Q, and Q*;
  • X 11 is selected from the group consisting of G, G*, A, A*, S, S*, P, P*, W, W*, Y, Y*, H, H*, T, T*, M, M*, N, N*, Q, and Q*; and
  • X 12 is selected from the group consisting of G, G*, A, A*, S, S*, P, P*, W, W*, Y, Y*, H, H*, T, T*, M, M*, N, N*, Q, and Q*;
  • G* is a non-natural derivative of G
  • A* is a non-natural derivative of A
  • S* is a non-natural derivative of S
  • P* is a non-natural derivative of P
  • W* is a non-natural derivative of W
  • Y* is a non-natural derivative of Y
  • H* is a non-natural derivative of H
  • T* is a non-natural derivative of T
  • M* is a non-natural derivative of M
  • N* is a non-natural derivative of N
  • Q* is a non-natural derivative of Q.
  • B is:
  • C is a polypeptide comprising from about 15 to about 25 amino acids, wherein the polypeptide comprises an amino acid sequence having at least 90% sequence identity to SEQ ID NO: 1.
  • X 2 is L or L*.
  • X 3 is P or P*. In some embodiments, X 4 is T or T*.
  • X 5 is K or K* .
  • X 6 is F or F*.
  • X 7 is R or R* .
  • X 8 is S or S*.
  • composition of Formula II is a composition of Formula
  • C is a polypeptide that binds HER3.
  • the present application further provides a pharmaceutical composition
  • composition comprising a composition provided herein, and a pharmaceutically acceptable carrier.
  • the present application further provides a method of imaging cancer in a subject, the method comprising:
  • compositions provided herein e.g., a composition of Formula I
  • the present application further provides a method of treating a cancer in a subject, the method comprising:
  • compositions provided herein e.g., a composition of Formula I
  • the present application further provides a method of monitoring treatment of a cancer in a subject, the method comprising:
  • compositions provided herein e.g., a composition of Formula I
  • the methods provided herein further comprise
  • the methods provided herein further comprise imaging the subject with a suitable imaging technique after step iii).
  • the therapeutic agent is a chemotherapeutic agent. In some embodiments, the therapeutic agent is a HER3 inhibitor. In some embodiments, the therapeutic agent is selected from the group consisting of patritumab, MM- 121 , U3-1402, GSK2849330, neratinib, lumretuzumab, U3-1287, Sym013, AV-203, trastuzumab, pertuzumab, cetrorelix, enzalutamide, and erlotinib.
  • the cancer is a solid tumor.
  • the cancer is selected from the group consisting of breast cancer, lung cancer, prostate cancer, gastric cancer, head and neck cancer, and ovarian cancer.
  • the prostate cancer is castration resistant prostate cancer.
  • the imaging technique is selected from the group consisting of magnetic resonance (MR) imaging, positron emission tomography (PET) imaging, fluorescent imaging, single photon emission computed tomography (SPECT), luminescent imaging, or any combination thereof.
  • MR magnetic resonance
  • PET positron emission tomography
  • SPECT single photon emission computed tomography
  • luminescent imaging or any combination thereof.
  • the imaging technique is positron emission tomography (PET) imaging.
  • Figure 1A shows a heat map of standardized phage supernatant binding to HER3 and BSA as a control. White indicates high binding and black indicates low binding.
  • Figure IB shows normalized binding of the 5 highest phage from supernatant phage ELISA, bars represent the mean of 6 replicates ⁇ SEM.
  • Figure 1C shows sequences of phages with high binding (1, 5, 17, 19, 26) and low binding (7, 22, 27) to HER3. ** P ⁇ 0.01.
  • Figure 2A shows peptide ELISA with increasing concentrations of HER3P1 demonstrates saturable binding and an affinity of 270 ⁇ 151 nM.
  • Triangles represent the mean of 6 replicates ⁇ SEM.
  • Figure 2B shows measured HER3P1 absorbance against two similar receptor tyrosine kinases and BSA, demonstrating approximately 4-fold selectivity over closely related proteins. Bars represent the mean of 6 replicates ⁇ SEM. * * P ⁇ 0.01.
  • Figure 3 A shows quantitative measurement ofHER3-l binding to moderate (MDA MB-453) and low (HCC-1954) HER3 expressing cells reveals significant differentiation by HER3P1 between cell types, in addition to significantly higher binding to a moderate HER3 cell line than a control peptide.
  • Figure 3B shows fluorescent microscopy visualizes peptide binding to MDA-MB- 453 cells, with little binding to HCC-1954 cells. * P ⁇ 0.05.
  • Figure 3C shows results of a 68 Ga-HER3Pl (i.e., 68 Ga-(NOTA-NCS)-HER3Pl) competitive binding assay (P ⁇ 0.05).
  • Figure 4A shows sagittal and axial maximum intensity projections of the radiolabeled peptide ( 68 Ga-(NOTA-NCS)-HER3Pl) in high HER3 expressing 22RV1 and low HER3 expressing HCC-1954 tumors. Peptide uptake is visualized in tumor and kidneys, which are the main route of clearance.
  • Figure 4B shows western blot analysis of HER3 and ⁇ -actin for tumors excised from mice following PET imaging.
  • Figure 4C shows quantification of HER3 : ⁇ -actin ratio for 22RV1 and HCC-1954 tumors. Bars represent the mean of 4 replicates ⁇ SEM.
  • Figure 4D shows linear regression with 95% confidence intervals for each tumor comparing PET TBR to ex vivo HER3:P-actin ratio. Linear significance was determined by a Pearson's correlation.** P ⁇ 0.01, *** p ⁇ 0.0001.
  • Figure 5A shows biodistnbution of 68 Ga-(NOTA-NCS)-HER3Pl in depicted organs as the mean injected dose per gram of tissue (%K)/g) of four 22RV1 tumor bearing mice ⁇ SD.
  • Figure 5B shows tumor to organ ratios calculated by biodistribution values from Figure 5A demonstrating specific accumulation in murine sites of non-tumor tissue HER3 expression.
  • Figure 5C shows linear regression of TBR from both biodistribution versus PET imaging. Significance of linearity determined by Pearson's correlation. * P ⁇ 0.05, *** P ⁇ 0.001.
  • Figure 6 shows mean phage binding normalized to parent (HER3P1) peptide.
  • Ml corresponds to mutant in position 1 following the cysteine, which is position 2 of the binding sequence.
  • Each of the subsequent phages (M2, M3, etc.) follow this naming convention. Bars represent the mean of 4 replicates with error bars denoting standard error measurements.
  • Figure 7 shows the measured SUVmean of mice bearing MDA-MB-453 HER2+ lapatinib resistant tumors were imaged pre-(circles) and post-therapy (squares) each point represents a matched pair of images from a single mouse imaged at day 0 pre-therapy and 2 days post therapy initiation. Bars represent mean and standard error measurement.
  • Figure 8 shows representative PET/MR images of a single mouse (mouse 1) pre- lapatinib (left image) and 2 days post-lapatinib therapy (right image). Tumors are circled and labeled with a white (T). Kidneys, which are the main route of peptide clearance, are labeled with a black K.
  • Molecular targeted cancer therapy while providing benefits for a number of indications, has so far failed to produce durable responses in a majority of patients (see e.g., Herbst et al, Journal of Clinical Oncology, 2005, 23:5892-5899). While no single mechanism is entirely responsible for targeted therapy resistance, a recurring theme is the upregulation of feedback loops that circumvent blockade of an oncogenic signaling pathway. Feedback loop signaling can be accomplished through alternative activation of a homologous pathway or through over-expression of a secondary oncogenic protein.
  • HER3 human epidermal growth factor receptor 3
  • HER3 human epidermal growth factor receptor 3
  • HER3 is a member of the epidermal growth factor (EGF) family of receptors that includes EGFR, HER2/ErbB2/neu, HER3/ErbB3 and HER4/ErbB4.
  • the EGFR family canonically function through ligand binding followed by dimerization, phosphorylation and downstream signaling through the MAPK and PI3K/AKT pathways (see e.g., Yarden,Y. Nature Reviews Molecular Cell Biology, 2001, 2: 127; Schlessinger, J. Cell, 2000, 103:211-225; and Ferguson et al, EMBO J. 2000, 19:4632-4643).
  • These receptors are major drivers of tumorigenesis, and multiple targeted therapies that block EGFR and HER2 are approved by the FDA (see e.g., Slamon et al, New England Journal of
  • HER3 has been less explored historically due to its mutated kinase domain, concurrent discoveries that its intracellular domain can in fact signal through transphosphorylation and that it is over- expressed in patients with resistance to a number of targeted therapies have brought it to the forefront of targeted therapy development (see e.g., Shi et al, Proceedings of the national Academy of Sciences, 2010, 107:7692-7697).
  • HER3-mediated therapeutic resistance may not be limited to therapies that target EGFR or HER2, as evidence has emerged to link HER3 with castration resistant prostate cancer as well.
  • NCT0213401S MM-121 (NCT00734305), U3-1402 (NCT02980341), and
  • GSK2849330 (NCT0196644S) are ongoing or have been completed recently, with limited success to date.
  • One limiting factor for the administration of such agents is identifying patient populations most likely to benefit from the therapy.
  • HER3 is particularly challenging to quantify by biopsy, because of its highly heterogeneous temporal and spatial expression.
  • PET imaging which provides a global and repeatable methodology to assess target expression, is highly compatible with HER3 expression diagnosis.
  • the present application provides a novel HER3 peptide for quantitative PET imaging of HER3 expressing tumors in murine models of multiple cancers.
  • the novel HER3 peptide, HER3P1 represents an accurate, pharmacokinetically favorable peptide imaging agent for HER3.
  • HER3 represents a critical protein in targeted therapies for a number of cancers and its highly dynamic expression requires an equally dynamic approach to quantification.
  • HER3 imaging with HER3P1 may represent a method to quantify HER3 and provide critical cellular feedback analysis.
  • A comprises an imaging agent
  • B is a linking group or a covalent bond
  • C is a polypeptide comprising from about 9 to about 75 amino acids, wherein the polypeptide comprises an amino acid sequence having at least 90% sequence identity to: NO: 13) wherein:
  • ⁇ ww refers to the bond between C and B
  • X 2 is selected from the group consisting of L, L*, I, I*, P, P*, V, V*, G, and G*;
  • X 3 is selected from the group consisting of P, P*, L, L*, I, I*, V, V*, G, and G*;
  • X 4 is selected from the group consisting of T, T*, S, S*, C, C*, M, and M*;
  • X 5 is selected from the group consisting of any L-amino acid, any D-amino acid, and any non-natural amino acid;
  • X 6 is selected from the group consisting of any L-amino acid, any D-amino acid, and any non-natural amino acid;
  • X 7 is selected from the group consisting of R, R*, H, H*, K, K*, N, N*, Q, and Q*, and
  • X 8 is selected from the group consisting of S, S*, T, T*, C, C*, M, and M*, wherein:
  • L* is a non-natural derivative of L
  • I* is a non-natural derivative of I
  • P* is a non-natural derivative of P
  • V* is a non-natural derivative of V
  • G* is a non-natural derivative of G
  • T* is a non-natural derivative of T
  • S* is a non-natural derivative of S
  • C* is a non-natural derivative of C
  • M* is a non- natural derivative of M
  • R* is a non- natural derivative of R
  • H* is a non-natural derivative of H
  • K* is a non-natural derivative of K
  • N* is a non-natural derivative of N
  • Q* is a non-natural derivative of Q.
  • A comprises an imaging agent selected from the group consisting of a paramagnetic ion, an x-ray imaging agent, a fluorophore, and a radioisotope.
  • A comprises a paramagnetic ion.
  • the paramagnetic ion is selected from the group consisting of chromium (III), manganese
  • A comprises an x-ray imaging agent.
  • the x-ray imaging agent is selected from the group consisting of lanthanum (III), gold (III), lead (II), bismuth (III), and iodinated x-ray imaging agents (e.g, diatrizoate, ioxaglate, metrizoate, iopamidol, iohexol, ioxilan, iopromide, iodixanol, and ioversol).
  • A comprises a fluorophore.
  • the fluorophore is selected from the group consisting of Alexa 350, Alexa 430, AMCA, BODIPY 630/650, BODIPY 650/665, BODIPY-FL, BODPY-R6G, 13BODLPY-TMR, BODLPY-TRX, cascade blue, Cy3, Cy5, 6-FAM, fluorescein isothiocyanate, HEX, 6- JOE, oregon green 488, oregon green 500, oregon green 514, quantum dots, pacific blue, REG, rhodamine green, rhodamine red, renographin, ROX, TAMRA, TET,
  • tetramethylrhodamine Texas Red
  • Alexafluor family Cy5, Cy5.5, Cy7, indocyanine green (ICG)
  • fluorescent proteins e.g., green fluorescent protein (GFP), red fluorescent protein (RFP), and dsRED.
  • A comprises a radioisotope.
  • the radioisotope provided herein is useful as an imaging agent in one or more of the methods provided herein.
  • the radioisotope is suitable for PET imaging.
  • the radioistope provided herein may also be useful in one or more therapeutic applications, for example, when administered to a subject in a therapeutically effective amount.
  • 13 l I and 64 Cu may be useful as imaging agents (e.g., as non-toxic and/or non-therapeutic radioisotopes) when administered to the subject at low concentrations (e.g., 5 mCi) and may also be useful as therapeutic agents (i.e., as toxic radioisotopes and/or therapeutic radioisotopes) when administered to the subject at a higher concentration.
  • the radioisotope is selected from the group consisting of 3 ⁇ 4, n C, 14 C, 18 F, 32 P, 35 S, 36 C1, 51 Cr, 52 Fe, 57 Co, 58 Co, 59 Fe, 64 Cu, 67 Cu, 67 Ga, 68 Ga, 75 Se, 76 Br, 77 Br, 89 Zr, 90 Y, 99m Tc, i n In, 123 I, 124 I, 125 I, 131 I, 152 Eu, 153 Sm, 166 Ho, 177 Lu, 186 Re, 188 Re, 201 T1, 203 Pb, 210 At, 211 At, 212 Bi, 213 Bi, and 225 Ac.
  • the radioisotope is selected from the group consisting of 3 H, n C, 14 C, 18 F, 35 S, 52 Fe, 58 Co, 64 Cu, 68 Ga, 76 Br, 77 Br, 89 Zr, m In, 123 I, 124 I, 125 I, 131 I, 186 Re, 188 Re, and 201 T1.
  • the radioisotope is 68 Ga.
  • A further comprises a chelating agent.
  • the chelating agent is selected from the group consisting of 1 ,4,7- triazacyclononanetriacetic acid (NOTA), 1,4,7,10-tetraazacyclododecane- 1,4,7, 10- tetraacetic acid (DOTA), 1 ,4,7-triazacyclononane-l-glutaric acid-4,7-diacetic acid (NODAGA), ethylene diamine tetra-acetic acid (EDTA), diethylene triaminepentaacetic acid (DTP A), cyclohexyl-l,2-diaminetetraacetic acid (CDTA), ethyl eneglycol-0,0'- bis(2-aminoethyl)-N,N,N',N'-tetraacetic acid (EGTA), N,N-bis(hydroxybenzyl)- ethylenediamine-N,N'-diacetic acid (HBED), triethylene tetramine hexaacetic acid
  • the chelating agent is selected from the group consisting of 1,4,7- triazacyclononanetriacetic acid (NOTA), 1,4,7,10-tetraazacyclododecane- 1,4,7, 10- tetraacetic acid (DOTA), 1,4,7-triazacyclononane-l-glutaric acid-4,7-diacetic acid (NODAGA), and 2,2',2"-(2-(4-isothiocyanatobenzyl)-l,4,7-triazonane-l,4,7-triyl)triacetic acid (NOTA-NCS).
  • NOTA 1,4,7- triazacyclononanetriacetic acid
  • DOTA 1,4,7,10-tetraazacyclododecane- 1,4,7, 10- tetraacetic acid
  • NODAGA 1,4,7-triazacyclononane-l-glutaric acid-4,7-diacetic acid
  • NCS 2,2',2"-(2-(4
  • the chelating agent is 1,4,7- triazacyclononanetriacetic acid (NOTA) or 2,2',2"-(2-(4-isothiocyanatobenzyl)- 1,4,7- triazonane-l,4,7-triyl)triacetic acid (NOTA-NCS).
  • NOTA-NCS 1,4,7- triazacyclononanetriacetic acid
  • the chelating agent is 2,2',2"-(2-(4-isothiocyanatobenzyl)-l,4,7-triazonane-l,4,7-triyl)triacetic acid (NOTA-NCS).
  • the chelating agent is:
  • B is a covalent bond. In some embodiments, B is a linking group.
  • B is a linking group comprising one or more Ci-30 alkyleneoxy groups, one or more independently amino acids, or any combination thereof.
  • B is a linking group comprising one or more Ci-30 alkyleneoxy groups. In some embodiments, B is a linking group comprising one or more -(OCH2CH2)- groups. In some embodiments, B is a linking group comprising one or more -(OCFhCFh groups, wherein p is an integer from 1 to 15, for example, 1 to 10, 1 to 5, 5 to 15, 5 to 10, or 10 to 15.
  • B is a linking group comprising a combination of one or more Ci-30 alkyleneoxy groups and one or more independently selected amino acids.
  • B is a linking group comprising one or more
  • B is a linking group comprising from about 1 to about 20 independently selected amino acids, for example, about 1 to about 15, about 1 to about 10, about 1 to about 5, about 5 to about 20, about 5 to about 15, about 5 to about 10, about 10 to about 20, about 10 to about 15, or about 15 to about 20 independently selected amino acids. In some embodiments, B is a linking group comprising from about 1 to about 10 independently selected amino acids.
  • B is a linking group comprising a sequence having at least 90% sequence identity to: -!- ⁇ 9. ⁇ 10. ⁇ 11. ⁇ 12 3 ⁇ 4
  • v ww refers to the bond between B and C
  • X 9 is selected from the group consisting of beta A, 6-aminohexnoic acid, 8- aminooctanoic acid, and 2-(2-(2-aminoethoxy)ethoxy)acetic acid;
  • X 10 is selected from the group consisting of G, G*, A, A*, S, S*, P, P*, W, W Y, Y*, H, H*, T, T*, M, M*, N, N*, Q, and Q*;
  • X 11 is selected from the group consisting of G, G*, A, A*, S, S*, P, P*, W, W Y, Y*, H, H*, T, T*, M, M*, N, N*, Q, and Q*; and
  • X 12 is selected from the group consisting of G, G*, A, A*, S, S*, P, P*, W, W Y, Y*, H, H*, T, T*, M, M*, N, N*, Q, and Q*;
  • G* is a non-natural derivative of G
  • A* is a non-natural derivative of A
  • S* is a non-natural derivative of S
  • P* is a non-natural derivative of P
  • W* is a non-natural derivative of W
  • Y* is a non-natural derivative of Y
  • H* is a non-natural derivative of H
  • T* is a non-natural derivative of T
  • M* is a non-natural derivative of M
  • N* is a non-natural derivative of N
  • Q* is a non-natural derivative of Q.
  • X 9 is beta A.
  • X 10 is G or G*. In some embodiments, X 10 is G. In some embodiments, X 11 is G or G*. In some embodiments, X 11 is G. In some embodiments, X 12 is G or G*. In some embodiments, X 12 is G. In some embodiments, at least two of X 10 , X 11 , and X 12 are G or G*. In some embodiments, at least two of X 10 , X 11 , and X 12 are G.
  • X 9 is beta A and at least two of X 10 , X 11 , and X 12 are G or G*. In some embodiments, X 9 is beta A and at least two of X 10 , X 11 , and X 12 are G.
  • each of X 10 , X 11 , and X 12 are G or G*. In some embodiments, each of X 10 , X 11 , and X 12 are G or G*. In some embodiments, each of X 10 , X 11 , and X 12 are G or G*. In some embodiments, each of X 10 , X 11 , and X 12 are G or G*. In some embodiments, each of X 10 , X 11 , and X 12 are G or G*.
  • each of X 10 , X 11 , and X 12 are G.
  • B is a linking group which is:
  • « ⁇ / refers to the bond between B and C.
  • C is a polypeptide that binds HER3.
  • C is a polypeptide comprising from about 9 to about 25 amino acids, wherein the polypeptide comprises an amino acid sequence having at least 90% sequence identity to SEQ ID NO: 13.
  • C is a polypeptide comprising from about 9 to about 25 amino acids, wherein the polypeptide comprises an amino acid sequence having at least 90% sequence identity
  • ⁇ ⁇ refers to a disulfide bond between the cysteine groups (i.e., a cyclized amino acid sequence formed by the disulfide bond between the cysteine groups).
  • X 2 is L or L*. In some embodiments, X 2 is L*. In some embodiments, X 2 is L.
  • X 3 is P or P*. In some embodiments, X 3 is P. In some embodiments, X 3 is P*.
  • X 4 is T or T*. In some embodiments, X 4 is T. In some embodiments, X 4 is T*. In some embodiments, X 5 is K or K* . In some embodiments, X 5 is K. In some embodiments, X 5 is K*.
  • X 6 is F or F*. In some embodiments, X 6 is F. In some embodiments, X 6 is F*.
  • X 7 is R or R*. In some embodiments, X 7 is R. In some embodiments, X 7 is R*.
  • X 8 is S or S*. In some embodiments, X 8 is S. In some embodiments, X 8 is S*.
  • X 2 is L or L*
  • X 3 is P or P*
  • X 4 is T or T*
  • X 6 is F or F*
  • X 7 is R or R*
  • X 8 is S or S*.
  • X 2 is L
  • X 3 is P
  • X 4 is T
  • X 5 is K
  • X 6 is F
  • X 7 is R
  • X 8 is S.
  • X 2 is L*
  • X 3 is P*
  • X 4 is T*
  • X 5 is K*
  • X 8 is S*.
  • composition of Formula I is a composition of Formula la:
  • X , X , X , X , X , and X are defined according to the definitions provided herein for compositions of Formula I, and wherein refers to a disulfide bond between the cysteine groups.
  • composition of Formula I is a composition of Formula lb:
  • A' is a radioisotope
  • X 2 , X 3 , X 4 , X 5 , X 6 , X 7 , and X 8 are defined according to the definitions provided herein for compositions of Formula I, and wherein refers to a disulfide bond between the cysteine groups.
  • composition of Formula I is a composition of Formula
  • compositions of Formula I wherein X , X , X , X , X , and X are defined according to the definitions provided herein for compositions of Formula I, and wherein refers to a disulfide bond between the cysteine groups.
  • composition of Formula I is:
  • composition of Formula I is a composition of Formula
  • X 2 , X 3 , X 4 , X 5 , X 6 , X 7 , and X 8 are defined according to the definitions provided herein for compositions of Formula I.
  • composition of Formula I is a composition of Formula lb:
  • A'-(NOTA-NCS)-beta A-G-G-G-C-X 2 -X 3 -X 4 -X 5 -X 6 -X 7 -X 8 -C SEQ m NQ .
  • A' is a radioisotope
  • X 2 , X 3 , X 4 , X 5 , X 6 , X 7 , and X 8 are defined according to the definitions provided herein for compositions of Formula I.
  • composition of Formula I is a composition of Formula
  • composition of Formula I is:
  • Ga-(NOTA-NCS)-beta A-G-G-G-C-L-P-T-K-F-R-S-C (SEQ ID NO: 17).
  • B comprises from about 1 to about 10 amino acids
  • compositions of Formula I are defined according to the definition provided herein for compositions of Formula I.
  • B comprises a sequence having at least 90% sequence identity to:
  • ⁇ svwv refers to the bond between B and C
  • X 9 is beta A.
  • X 10 is G or G*. In some embodiments, X 10 is G.
  • X 11 is G or G*. In some embodiments, X 11 is G.
  • X 12 is G or G*. In some embodiments, X 12 is G.
  • At least two of X 10 , X 11 , and X 12 are G or G*. In some embodiments, at least two of X 10 , X 11 , and X 12 are G.
  • X 9 is beta A and at least two of X 10 , X 11 , and X 12 are G or G*. In some embodiments, X 9 is beta A and at least two of X 10 , X 11 , and X 12 are G.
  • each of X 10 , X 11 , and X 12 are G or G*. In some embodiments, each of X 10 , X 11 , and X 12 are G.
  • B is: beta A-G-G-G-1 ⁇ 2-
  • ⁇ *wv refers to the bond between B and C.
  • C is a polypeptide comprising from about 9 to about 25 amino acids, wherein the polypeptide comprises an amino acid sequence having at least 90% sequence identity to SEQ ID NO: 13. In some embodiments, C is a polypeptide comprising from about 9 to about 25 amino acids, wherein the polypeptide comprises an amino acid sequence having at least 90% sequence identity to SEQ ID NO: 12. In some embodiments, C is a polypeptide that binds HER3.
  • X 2 is L or L*. In some embodiments, X 2 is L*. In some embodiments, X 2 is L.
  • X 3 is P or P*. In some embodiments, X 3 is P. In some embodiments, X 3 is P*.
  • X 4 is T or T*. In some embodiments, X 4 is T. In some embodiments, X 4 is T*.
  • X 5 is K or K* . In some embodiments, X 5 is K. In some embodiments, X 5 is K*.
  • X 6 is F or F*. In some embodiments, X 6 is F. In some embodiments, X 6 is F*.
  • X 7 is R or R*. In some embodiments, X 7 is R. In some embodiments, X 7 is R*.
  • X 8 is S or S*. In some embodiments, X 8 is S. In some embodiments, X 8 is S*.
  • X 2 is L or L*
  • X 3 is P or P*
  • X 4 is T or T*
  • X 6 is F or F*
  • X 7 is R or R*; and X 8 is S or S*.
  • X 2 is L
  • X 3 is P
  • X 4 is T
  • X 5 is K
  • X 6 is F
  • X 7 is R
  • X 8 is S.
  • X 2 is L*
  • X 3 is P*
  • X 4 is T*
  • X 5 is K*
  • X 6 is F*
  • X 7 is R*
  • X 8 is S*.
  • composition of Formula II is a composition of Formula
  • X , X , X , X , X , and X are defined according to the definitions provided herein for compositions of Formula II, and wherein refers to a disulfide bond between the cysteine groups.
  • the composition of Formula II is a composition of Formula lib: beta A-G-G-G-C-X 2 -X 3 -X 4 -X 5 -X 6 -X 7 -X 8 -C (SEQ ID N o 1 7) lib
  • X 2 , X 3 , X 4 , X 5 , X 6 , X 7 , and X 8 are defined according to the definitions provided herein for compositions of Formula II.
  • composition of Formula II is:
  • composition of Formula II is:
  • ⁇ ⁇ refers to a disulfide bond between the cysteine groups.
  • the present application further provides a polypeptide suitable for binding HER3.
  • the polypeptide comprises from about 9 to about 75 amino acids.
  • the polypeptide comprises an amino acid sequence having at least one amino acid sequence having at least one amino acid sequence having at least one amino acid sequence having at least one amino acid sequence having at least one amino acid sequence having at least one amino acid sequence having at least one amino acid sequence having at least one amino acid sequence having at least one amino acid sequence having at least one amino acid sequence having at least one amino acid sequence having at least one amino acid sequence having at least one amino acid sequence having at least one amino acid sequence having at least one amino acid sequence having at least
  • the polypeptide comprises from about 15 to about 25 amino acids, wherein the polypeptide comprises an amino acid sequence having at least 90% sequence identity to SEQ ID NO: 19. In some embodiments, the polypeptide comprises from about 15 to about 25 amino acids, wherein the polypeptide comprises an amino acid sequence having at least 90% sequence identity to SEQ ID NO: 20:
  • X 2 is L or L*. In some embodiments, X 2 is L*. In some embodiments, X 2 is L.
  • X 3 is P or P*. In some embodiments, X 3 is P. In some embodiments, X 3 is P*.
  • X 4 is T or T*. In some embodiments, X 4 is T. In some embodiments, X 4 is T*.
  • X 5 is K or K*. In some embodiments, X 5 is K. In some embodiments, X 5 is K*.
  • X 6 is F or F*. In some embodiments, X 6 is F. In some embodiments, X 6 is F*.
  • X 7 is R or R*. In some embodiments, X 7 is R. In some embodiments, X 7 is R*.
  • X 8 is S or S*. In some embodiments, X 8 is S. In some embodiments, X 8 is S*.
  • X 2 is L or L*
  • X 3 is P or P*
  • X 4 is T or T*
  • X 6 is F or F*
  • X 7 is R or R*
  • X 8 is S or S*.
  • X 2 is L
  • X 3 is P
  • X 4 is T
  • X 5 is K
  • X 6 is F
  • X 7 is R
  • X 8 is S. In some embodiments:
  • X 2 is L*
  • X 3 is P*
  • X 4 is T*
  • X 5 is K*
  • X 6 is F*
  • X 7 is R*
  • X 8 is S*.
  • polypeptide is:
  • polypeptide is:
  • refers to a disulfide bond between the cysteine groups.
  • compositions provided herein can be prepared, for example, according to the procedures described below.
  • compositions provided herein comprising polypeptides can be prepared, for example, using standard techniques for the preparation of peptide bonds ⁇ e.g., solid-phase synthetic techniques as described in Merrifield et al, Journal of the American Chemical Society 85.14 (1963): 2149-2154).
  • Peptide synthetic techniques are well known to those of skill in the art and are described, for example, in Bodanszky et al, Gastroenterology 71 (1976): 965-970; Houghten, Proceedings of the National Academy of Sciences 82.15 (1985): 5131-5135; Stewart et al, Solid phase peptide synthesis. Pierce Chemical Company, 1984.
  • either the amino or carboxyl group of the first amino acid residue is protected by a suitable, selectively removable protecting group.
  • a different, selectively removable protecting group is utilized for amino acids containing a reactive side group, such as lysine.
  • polypeptides of the present application may be prepared using solid phase synthetic techniques.
  • the amino acid is attached to an inert solid support through its unprotected carboxyl or amino group.
  • the protecting group of the amino or carboxyl group is then selectively removed and the next amino acid in the sequence having the complementary (e.g., amino or carboxyl) group suitably protected is admixed and reacted with the residue already attached to the solid support.
  • the protecting group of the amino or carboxyl group is then removed from the newly added amino acid residue, and a further amino acid (e.g. an appropriately protected amino acid) is then added. This procedure may be repeated until the desired polypeptide length has been prepared.
  • any remaining terminal and side group protecting groups (and solid support) can be removed sequentially or concurrently to provide the final peptide. Additional reactions may be necessary, as described elsewhere, to form intramolecular linkages to restrain
  • Bifunctional cross-linking reagents have been extensively used the preparation of affinity matrices, modification and stabilization of diverse structures, identification of ligand and receptor binding sites, and structural studies.
  • Homobifunctional reagents having two identical functional groups have been shown to be highly efficient in cross- linking identical and different polypeptides or residues of a polypeptide, and the linking of polypeptide ligands to their specific binding sites.
  • Heterobifunctional reagents contain two different functional groups. By taking advantage of differential reactivity of the different functional groups, cross-linking can be controlled both selectively and sequentially.
  • bifunctional cross-linking reagents can be divided according to the specificity of their functional groups, including, but not limited to, amino, sulfhydryl, guanidino, indole, carboxyl specific groups.
  • Many heterobifunctional cross- linking reagents contain a primary amine-reactive group and a thiol-reactive group.
  • heterobifunctional cross-linking reagents and methods of using the cross-linking reagents are described in U. S. Patent 5,889,155, the disclosure of which is incorporated herein by reference in its entirety.
  • the cross-linking reagents combine a nucleophilic hydrazide residue with an electrophilic maleimide residue, allowing coupling in one example, of aldehydes to free thiols.
  • the cross-linking reagent can be modified to cross-link various functional groups and is thus useful for cross- linking polypeptides.
  • Table 1 details certain hetero-bifunctional cross-linkers considered useful for preparing compositions comprising a group B described herein (e.g., a linking group B as described herein).
  • compositions where a particular peptide does not contain a residue amenable for a particular cross-linking reagent in its native sequence conservative genetic or synthetic amino acid changes in the primary sequence can be utilized.
  • imaging agents are known in the art, as are methods for their attachment to antibodies (see, for e.g., U.S. Patents 5,021,236; 4,938,948; and 4,472,509, the disclosure of each of which is incorporated herein by reference in its entirety).
  • Radioactively labeled compositions provided herein may be prepared according to well- known methods in the art.
  • monoclonal antibodies can be iodinated by contact with sodium and/or potassium iodide and a chemical oxidizing agent such as sodium hypochlorite, or an enzymatic oxidizing agent, such as lactoperoxidase.
  • compounds of Formula I provided herein may be labeled with 68 Ga by radiometalation of a bifunctional chelator provided herein (e.g., NOTA, DOTA,
  • NODAGA NODAGA, or NOTA-NCS
  • NODAGA NODAGA, or NOTA-NCS
  • the reactions for preparing compositions described herein can be carried out in suitable solvents which can be readily selected by one of skill in the art of organic synthesis.
  • suitable solvents can be substantially non-reactive with the starting materials (reactants), the intermediates, or products at the temperatures at which the reactions are carried out, (e.g., temperatures which can range from the solvent's freezing temperature to the solvent's boiling temperature).
  • a given reaction can be carried out in one solvent or a mixture of more than one solvent.
  • suitable solvents for a particular reaction step can be selected by the skilled artisan.
  • compositions described herein can involve the protection and deprotection of various chemical groups.
  • the need for protection and deprotection, and the selection of appropriate protecting groups, can be readily determined by one skilled in the art.
  • the chemistry of protecting groups can be found, for example, in T. W. Greene and P. G. M. Wuts, Protective Groups in Organic Synthesis, 3 rd Ed., Wiley & Sons, Inc., New York (1999).
  • Reactions can be monitored according to any suitable method known in the art.
  • product formation can be monitored by spectroscopic means, such as nuclear magnetic resonance spectroscopy (e.g., 3 ⁇ 4 or 13 C), infrared spectroscopy, spectrophotometry (e.g., UV- visible), mass spectrometry, or by chromatographic methods such as high performance liquid chromatography (HPLC), liquid
  • LCMS chromatography-mass spectroscopy
  • TLC thin layer chromatography
  • compositions can be purified by those skilled in the art by a variety of methods, including high performance liquid chromatography (HPLC) and normal phase silica chromatography.
  • HPLC high performance liquid chromatography
  • HPLC normal phase silica chromatography
  • each divalent linking substituent include both the forward and backward forms of the linking substituent.
  • -NR(CR'R") n - includes both -NR(CR'R") n - and -(CR'R") n NR-.
  • the Markush variables listed for that group are understood to be linking groups.
  • Tautomeric forms result from the swapping of a single bond with an adjacent double bond together with the concomitant migration of a proton.
  • Tautomeric forms include prototropic tautomers which are isomeric protonation states having the same empirical formula and total charge.
  • Example prototropic tautomers include ketone - enol pairs, amide - imidic acid pairs, lactam - lactim pairs, enamine - imine pairs, and annular forms where a proton can occupy two or more positions of a heterocyclic system, for example, 1H- and 3H- imidazole, 1H-, 2H- and 4H- 1 ,2,4-triazole, 1H- and 2H- isoindole, and 1H- and 2H- pyrazole.
  • Tautomeric forms can be in equilibrium or sterically locked into one form by appropriate substitution.
  • compounds and compositions provided herein can also include all isotopes of atoms occurring in the intermediates or final compounds.
  • Isotopes include those atoms having the same atomic number but different mass numbers.
  • an atom is designated as an isotope or radioisotope (e.g., deuterium, [ n C], [ 18 F])
  • the atom is understood to comprise the isotope or radioisotope in an amount at least greater than the natural abundance of the isotope or radioisotope.
  • an atom is designated as "D” or "deuterium”
  • the position is understood to have deuterium at an abundance that is at least 3000 times greater than the natural abundance of deuterium, which is 0.015% (i.e., at least 45% incorporation of deuterium).
  • preparation of compounds and compositions described herein can involve the addition of acids or bases to affect, for example, catalysis of a desired reaction or formation of salt forms such as acid addition salts.
  • Example acids can be inorganic or organic acids and include, but are not limited to, strong and weak acids.
  • Some example acids include hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, /7-toluenesulfonic acid, 4-nitrobenzoic acid, methanesulfonic acid, benzenesulfonic acid, trifluoroacetic acid, and nitric acid.
  • Some weak acids include, but are not limited to acetic acid, propionic acid, butanoic acid, benzoic acid, tartaric acid, pentanoic acid, hexanoic acid, heptanoic acid, octanoic acid, nonanoic acid, and decanoic acid.
  • Example bases include lithium hydroxide, sodium hydroxide, potassium hydroxide, lithium carbonate, sodium carbonate, potassium carbonate, and sodium bicarbonate.
  • Some example strong bases include, but are not limited to, hydroxide, alkoxides, metal amides, metal hydrides, metal dialkylamides and arylamines, wherein; alkoxides include lithium, sodium and potassium salts of methyl, ethyl and t-butyl oxides; metal amides include sodium amide, potassium amide and lithium amide; metal hydrides include sodium hydride, potassium hydride and lithium hydride; and metal dialkylamides include lithium, sodium, and potassium salts of methyl, ethyl, n-propyl, z ' so-propyl, n- butyl, tert-butyl, trimethylsilyl and cyclohexyl substituted amides.
  • the compounds (e.g., polypeptides) and compositions provided herein are substantially isolated.
  • substantially isolated is meant that the compound is at least partially or substantially separated from the environment in which it was formed or detected.
  • Partial separation can include, for example, a composition enriched in the compounds provided herein.
  • Substantial separation can include compositions containing at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, at least about 97%, or at least about 99% by weight of the compounds provided herein, or salt thereof. Methods for isolating compounds and compositions are routine in the art.
  • phrases "pharmaceutically acceptable” is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • non-natural amino acid refers to any derivative of a natural amino acid including D forms, and ⁇ and ⁇ amino acid derivatives. It is noted that certain amino acids, e.g., hydroxyproline, that are classified as a non- natural amino acid herein, may be found in nature within a certain organism or a particular protein.
  • non-natural amino acids or amino acid derivatives may be constituents of the compositions described herein (common abbreviations are in parentheses): ⁇ - Alanine (beta A, ⁇ -Ala, ⁇ , or ⁇ - ⁇ ), ⁇ -Aminobutyric Acid (GAB A), 2- Aminobutyric Acid (2- Abu), a ⁇ -Dehydro-2-aminobutyric Acid ( ⁇ -Abu), 1- Aminocyclopropane- 1 -carboxylic Acid (ACPC), Aminoisobutyric Acid (Aib), 2-Amino- thiazoline-4-carboxylic Acid, 5-Aminovaleric Acid (5-Ava), 6-Aminohexanoic Acid (6- Ahx), 8-Aminooctanoic Acid (8-Aoc), 11 - Aminoundecanoic Acid (11-Aun), 12- Aminododecanoic Acid (12-Ad
  • Cyclohexylglycine (Chg), 2,3-Diaminopropionic Acid (Dpr), 2,4-Diaminobutyric Acid (Dbu), 3,4-Dichlorophenylalanine (3,4-C12-Phe), 3,4-Diflurophenylalanine (3,4-F2-Phe), 3,5-Diiodotyrosine (3,5-I2-Tyr), ori zo-Fluorophenylalanine (2-F-Phe), meta- Fluorophenylalanine (3-F-Phe), />ara-Fluorophenylalanine (4-F-Phe), «3 ⁇ 4eto-fluorotyrosine (3-10 F-Tyr), Homoserine (Hse), Homophenylalanine (Hfe), Homotyrosine (Htyr), 5- Hydroxytryptophan (5-OH-Trp), Hydroxyproline (Hyp), /?ara-Iod
  • Octahydroindole-2-carboxylic Acid Oic
  • Penicillamine Pen
  • Pentafluorophenylalanine F5-Phe
  • Phenylglycine Phg
  • Pipecolic Acid Pip
  • Propargylglycine Pra
  • the present application further provides methods of imaging HER3.
  • the method of imaging is performed in a cell, a tissue, a cell sample, a tissue sample, or a subject.
  • the term "subject,” refers to any animal, including mammals and invertebrates. For example, mice, rats, other rodents, rabbits, dogs, cats, swine, cattle, sheep, horses, primates, fish, and humans. In some
  • the subject is a human. In some embodiments, the subject is a mouse. In some embodiments, the method comprises administering to the subject an effective amount of a composition provided herein (e.g., a composition of Formula I). In some embodiments, the method is an in vitro method. In some embodiments, the method is an in vivo method.
  • a composition provided herein e.g., a composition of Formula I
  • the method is an in vitro method. In some embodiments, the method is an in vivo method.
  • the present application provides a method of imaging cancer in a subject, the method comprising:
  • compositions provided herein ⁇ e.g., a composition of Formula I
  • the present application further provides a method of treating a cancer in a subject, the method comprising:
  • compositions provided herein ⁇ e.g., a composition of Formula I);
  • the present application further provides a method of monitoring treatment of a cancer in a subject, the method comprising: i) administering to the subject an effective amount of a composition provided herein (e.g., a composition of Formula I);
  • the methods provided herein further comprise
  • the methods provided herein further comprise imaging the subject with a suitable imaging technique after step iii).
  • the methods provided herein further comprise waiting a time sufficient to allow the compound to accumulate at a cell or tissue site (e.g., a cell or tissue site in a subject) associated with the disease, prior to imaging.
  • a cell or tissue site e.g., a cell or tissue site in a subject
  • the methods provided herein further comprise waiting a time sufficient to allow the compound to bind HER3 at a cell or tissue site (e.g., a cell or tissue site in a subject) associated with the cancer, prior to imaging.
  • the time sufficient is from about 30 seconds to about 24 hours, for example, about 30 seconds to about 24 hours, about 30 seconds to about 12 hours, about 30 seconds to about 6 hours, about 30 seconds to about 2 hours, about 30 seconds to about 1 hour, about 30 seconds to about 30 minutes, about 30 seconds to about 10 minutes, about 10 minutes to about 24 hours, about 10 minutes to about 12 hours, about 10 minutes to about 6 hours, about 10 minutes to about 2 hours, about 10 minutes to about 1 hour, about 10 minutes to about 30 minutes, about 30 minutes to about 24 hours, about 30 minutes to about 12 hours, about 30 minutes to about 6 hours, about 30 minutes to about 2 hours, about 30 minutes to about 1 hour, about 1 hour to about 24 hours, about 1 hour to about 12 hours, about 1 hour to about 6 hours, about 1 hour to about 2 hours,
  • the cancer is a solid tumor. In some embodiments, the cancer is selected from the group consisting of breast cancer, lung cancer, prostate cancer, gastric cancer, head and neck cancer, and ovarian cancer. In some embodiments, the cancer is selected from the group consisting of breast cancer, lung cancer, and prostate cancer. In some embodiments, the cancer is prostate cancer. In some embodiments,
  • the prostate cancer is castration resistant prostate cancer.
  • the cancer is breast cancer.
  • the imaging technique is selected from the group consisting of magnetic resonance (MR) imaging, positron emission tomography (PET) imaging, fluorescent imaging, single photon emission computed tomography (SPECT), luminescent imaging, or any combination thereof.
  • the imaging technique is selected from the group consisting of magnetic resonance (MR) imaging, positron emission tomography (PET) imaging, or a combination thereof.
  • the imaging technique is positron emission tomography (PET) imaging.
  • the phrase "therapeutically effective amount” refers to the amount of active compound or pharmaceutical agent that elicits the biological or medicinal response that is being sought in a tissue, system, animal, individual or human by a researcher, veterinarian, medical doctor or other clinician.
  • treating refers to one or more of (1) inhibiting the disease; for example, inhibiting a disease, condition or disorder in an individual who is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e., arresting further development of the pathology and/or symptomatology); and (2) ameliorating the disease; for example, ameliorating a disease, condition or disorder in an individual who is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e., reversing the pathology and/or symptomatology) such as decreasing the severity of disease or reducing or alleviating one or more symptoms of the disease.
  • compositions provided herein can be administered in combination with one or more of the additional therapeutic agents provided herein.
  • additional therapeutic agents include, but are not limited to, anesthetic agents (e.g., for use in combination with a surgical procedure), chemotherapeutic agents, and therapeutic antibodies.
  • the additional therapeutic agent is an anesthetic agent.
  • exemplary anesthetic agents include, but are not limited to, local anesthetics (e.g., lidocaine, procain, ropivacaine) and general anesthetics (e.g., desflurane, enflurane, halothane, isoflurane, methoxyflurane, nitrous oxide, sevoflurane, mmobarbital, methohexital, thiamylal, thiopental, diazepam, lorazepam, midazolam, etomidate, ketamine, propofol, alfentanil, fentanyl, remifentanil, buprenorphine, butorphanol, hydromorphone levorphanol, meperidine, methadone, morphine, nalbuphine,
  • local anesthetics e.g., lidocaine, procain,
  • the additional therapeutic agent is a chemotherapeutic agent.
  • chemotherapeutic agents include, but are not limited to, cisplatin, doxorubicin, taxol, etoposide, irinotecan, topotecan, paclitaxel, docetaxel, epothilones, tamoxifen, 5-fluorouracil, methotrexate, temozolomide, cyclophosphamide, tipifarnib, gefitinib, erlotinib, imatinib, gemcitabine, uracil mustard, chlormethine, ifosfamide, melphalan, chlorambucil, pipobroman, triethylenemelamine, busulfan, carmustine, lomustine, streptozocin, dacarbazine, floxuridine, cytarabine, 6-mercaptopurine, 6- thioguanine, fludarabine phosphate, ox
  • Exemplary therapeutic antibodies include, but are not limited to, patritumab, lumretuzumab, trastuzumab, pertuzumab, MM-121, U3-1402, AV-203, and
  • the therapeutic agent is a HER3 inhibitor.
  • the therapeutic agent is selected from the group consisting of patritumab, MM-121, U3-1402, GSK2849330, neratinib, lumretuzumab, U3-1287, Sym013, AV-203, and erlotinib.
  • the therapeutic agent is a HER2 inhibitor.
  • the HER2 inhibitor is selected from the group consisting of lapatinib, trastuzumab, pertuzumab, and erlotinib.
  • the therapeutic agent is a PI3K inhibitor.
  • the PI3K inhibitor is GDC-0941.
  • the therapeutic agent is a AKT inhibitor.
  • the AKT inhibitor is GDC-0068.
  • the therapeutic agent is an androgen receptor antagonist.
  • the androgen receptor antagonist is enzalutamide.
  • the therapeutic agent is a gonadotrophin-releasing hormone antagonist.
  • the gonadotrophin-releasing hormone antagonist is cetrorelix.
  • the additional therapeutic agent is administered simultaneously with a composition provided herein. In some embodiments, the additional therapeutic agent is administered after administration of the composition provided herein. In some embodiments, the additional therapeutic agent is administered prior to administration of the composition herein. In some embodiments, the composition provided herein is administered during a surgical procedure. In some embodiments, the composition provided herein is administered in combination with an additional therapeutic agent during a surgical procedure.
  • the additional therapeutic agents provided herein can be effective over a wide dosage range and are generally administered in an effective amount. It will be understood, however, that the amount of the therapeutic agent actually administered will usually be determined by a physician, according to the relevant circumstances, including the condition to be imaged, the chosen route of administration, the actual compound administered, the age, weight, and response of the individual subject, the severity of the subject's symptoms, and the like.
  • compositions and therapeutic agents provided herein can be administered in the form of pharmaceutical formulations. These formulations can be prepared as described herein or elsewhere, and can be administered by a variety of routes, depending upon whether local or systemic treatment is desired and upon the area to be treated.
  • the administration is selected from the group consisting of pulmonary administration (e.g., by inhalation or insufflation of powders or aerosols, including by nebulizer, intratracheal administration, or intranasal administration), oral administration, or parenteral administration (e.g., intravenous, intraarterial, subcutaneous, intraperitoneal, intramuscular or injection or infusion, intracranial, intrathecal, intraventricular administration, and the like).
  • the administration is intravenous or nasal administration.
  • Parenteral administration can be in the form of a single bolus dose, or may be, for example, by a continuous perfusion pump.
  • Conventional pharmaceutical carriers, aqueous, powder, or oily bases, thickeners and the like, may be necessary or desirable.
  • compositions which contain, as the active ingredient, a composition provided herein in combination with one or more
  • the nanoparticle composition may be, for example, mixed with an excipient or diluted by an excipient.
  • the excipient serves as a diluent, it can be a solid, semi-solid, or liquid material, which acts as a vehicle, carrier, or medium for the nanoparticle composition.
  • the pharmaceutical formulations can be in the form of powders, lozenges, elixirs, suspensions, emulsions, solutions, syrups, aerosols (as a solid or in a liquid medium), sterile injectable solutions, sterile packaged powders, and the like.
  • MDA-MB-453, HCC1954 and 22Rvl cells were purchased from American Type Culture Collection (Manassas, VA) and cultured in RPMI-1640 media supplemented with 10% fetal bovine serum and 1 % penicillin and streptomycin.
  • RPMI-1640 media supplemented with 10% fetal bovine serum and 1 % penicillin and streptomycin.
  • a one-way ANOVA with a Dunnett test to correct for multiple comparisons was performed.
  • a sigmoidal dose response curve was fit to the peptide affinity data using Graphpad Prism 6.0.
  • a cysteine-constrained randomized 7-mer library (New England Biolabs, Ipswich, MA) was utilized for the phage display selection.
  • HER3 extracellular domain (ECD) (R&D Systems, Minneapolis, MN) was conjugated with long chain biotin (Thermo Scientific, Waltham, MA) utilizing standard NHS ester chemistry and purified by size exclusion chromatography with a 10 kDa molecular weight cut-off column (Genesee Scientific, San Diego, CA ).
  • HER3 ECD and bovine serum albumin as a control were adsorbed to Nunc Maxisorp 96 well plates (Sigma, St. Louis, MO).
  • the relative binding was compared to a control phage bearing no peptide and represented by the heat map in Figure 1 A.
  • the phages with 5 best binding ratios of HER3 to BSA were chosen for amplification and purification to ensure no other supernatant factor was obscuring the signal.
  • ELISA was performed in the exact same manner and the absorbance of each phage to either HER3 or BSA was quantified by spectrophotometer. Phagemid DNA from each of the five phages in addition to three phages with no binding per the initial ELISA were recovered by plasmid miniprep isolation and automated Sanger DNA sequencing was performed by the CCIB DNA Core Facility at Massachusetts General Hospital
  • all 5 were sequenced to ascertain the amino acid composition of their displayed peptide.
  • each of the sequenced phage displayed an identical peptide.
  • three phage which did not demonstrate HER3 specific binding were also sequenced, and each had a different peptide sequence than the convergent sequence, as shown in Figure 1C.
  • the peptide sequence was covalently linked to a biotin-conjugated N-terminal tri-glycine linker using standard Fmoc chemistry. The purity and molecular weight were determined by high performance liquid chromatography and mass spectrometry. In order to confirm HER3 affinity, the peptide was analyzed for binding to HER3 ECD immobilized to Nunc Maxisorp 96 well microtiter plates. Following blocking with 1% non-fat dry milk, increasing concentrations of peptide were incubated with target protein for 1 h at 37°C.
  • HER3P1 bound in a sigmoidal manner, with an affinity of 270 ⁇ 151 nM, as shown in Figure 2A.
  • the absorbance of HER3P1 binding to HER3 was 0.82 ⁇ 0.03, whereas binding to EGFR was 0.21 ⁇ 0.09, HER2 was 0.22 ⁇ 0.07 and BSA was 0.15 ⁇ 0.01 (P ⁇ 0.001 for all), indicating that the peptide was highly specific for HER3 and suitable for cell binding analysis, as shown in Figure 2B.
  • MDA-MB-453 breast cancer cells which express moderate levels of HER3 and HCC-1954 cells which express low levels of HER3, were chosen to assess the ability of the peptide to discriminate between relatively close expression levels in vitro.
  • Cells were seeded at a density of lxl 0 5 cells/well in 96 well plates (Fisher) and grown overnight for 24 h in fetal bovine serum supplemented medium.
  • HER3 peptide or a control peptide were added at 250 nM in media and incubated for 1 h at 37°C. Cells were washed 3x with PBS and bound peptide detected by addition of streptavidin-HRP.
  • MDA-MB-453 and HCC-1954 cells were fixed in 10% formalin and dried onto microscope slides overnight. Following rehydration with TBS, slides were blocked with 2% BSA in TBS for 1 h prior to addition of peptide at a concentration of 250 nM in 0.1 % TBST and incubation at room temperature for 1 h. Cells were washed 3x with 0.1% TBST and neutravidin-Alexafluor488 was added to cells and incubated at room temperature for 1 h. Cells were washed, mounted with Vectorshield mounting media with DAPI (Vector Labs, Burlingame, CA) and visualized by inverted microsope (Olympus, Tokyo, Japan).
  • DAPI Vector Labs, Burlingame, CA
  • HER3P1 Cell binding for HER3P1 was quantified and absorbance for MDA-MB-453 was 0.29 ⁇ 0.06, whereas it was significantly lower for HCC-1954 cells 0.13 ⁇ 0.06 (P ⁇ 0.05). Additionally HER3P1 binding was significantly higher to MDA-MB-453 cells than the control peptide (0.12 ⁇ 0.06, P ⁇ 0.05), whereas there was no difference between the control peptide and the HER3P1 for binding to HCC-1954 cells, as shown in Figure 3 A. The strong selectivity of the peptide was also apparent using fluorescent microscopy, with high binding to MDA-MB-453 cells and almost no visualization of HCC-1954 cells, as shown in Figure 3B.
  • a NOTA-NCS conjugated HER3 peptide was synthesized in the same manner as the biotinylated peptide, with NOTA-NCS being substituted for biotin.
  • the peptide was radio labeled with the radiometal 68 Ga eluted from a 68 Ge/ 68 Ga generator (iThemba, South Africa) in 0.05M HC1.
  • Peptide was purified from free 68 Ga by reverse phase CI 8 cartridge (Waters, Milford, MA) and purity determined by ITLC. The peptide was then prepared in normal saline to a specific activity of approximately 300 MBq/mg for injection into mice. Nu/nu mice bearing either 22RV1 (high HER3 expressing prostate cancer) or HCC-1954 (low HER3 expressing breast cancer) tumors were implanted in the right upper flank of mice and grown to approximately 5-7 mm. Radiolabeled HER3 peptide was injected intravenously and allowed to circulate for 1 h prior to PET imaging. PET images were acquired on an Inveon micro PET /CT (Siemens, Knoxville, TN) for 15 minutes in list mode, followed by CT acquisition.
  • CT Inveon micro PET /CT
  • the tumor and blood uptake was calculated in a 3D region of interest drawn around the tumor and heart, respectively, using CT guidance. Images were post-processed using VivoQuant (InviCRO, Boston, MA).
  • HER3 sc-81455, Santa Cruz Biotech, Dallas, TX
  • ⁇ -actin 13E5, Cell Signaling, Danvers, MA
  • PET imaging demonstrated high 68 Ga-(NOTA-NCS)-HER3Pl tumor uptake in 22RV1 tumor bearing mice, with much lower levels in HCC-1954 tumor bearing mice, as shown in Figure 4A.
  • Off -target peptide accumulation was minimal, with uptake in the kidneys and bladder consistent with the normal route of peptide clearance.
  • HER3 and ⁇ -actin were quantified by Western blot, as shown in Figure 4B.
  • This data confirmed that the HER3P1 uptake was highly correlated with HER3 expression and accurately quantified total HER3 in vivo, as shown in Figure 4C. Additionally, biodistribution analysis of HER3P1 was performed.
  • Sites of accumulation included the HER3+ tumors (0.50 ⁇ 0.18%K)/g) and HER3+ organs such as the stomach (0.30 ⁇ 0.07%K)/g), intestines (0.48 ⁇ 0.15%K)/g) and lungs (0.70 ⁇ 0.14%ID/g), in addition to kidneys (10.1 ⁇ 1.67%ID/g) as a route of clearance, as shown in Figure 5 A.
  • HER3P1 demonstrated excellent tumor to background ratios (1.59-3.32) in HER3+ tumors which were significantly higher than the low HER3 expressing control tumors (0.84-0.93). Furthermore, ex vivo analysis of imaged tumors revealed a high (P ⁇ 0.001) correlation between HER3 peptide uptake and both HER3 protein expression and percent injected dose per gram. These results indicate that HER3P1 represents a promising, clinically translatable HER3 imaging agent, and future translational efforts are being sought. PET imaging also revealed high tumor to blood ratios in the HER3 positive 22RV1 cell line, with background levels in HER3 -negative HCC-1954 tumors.
  • ex vivo analysis of the tumors used for PET imaging provided a highly significant correlation between protein expression and PET TBR, indicating an accurate and robust method of HER3 quantification suitable for exploration in both pre-clinical and clinical trials.
  • a competitive binding assay was also performed according to the following procedures. Ten ⁇ of purified 68 Ga-HER3Pl was added to lxlO 5 MDA-MB-453 cells and incubated for 1 h. After incubation, cells were washed 3X with TBS and bound radioactivity quantified by a Wallac gamma counter (Perkin Elmer, Waltham, MA). As a control, excess (100 ⁇ ) peptide was added to cells and bound peptide measured in the same manner. The 68 Ga-HER3Pl bound to MDA-MB-453 cells and binding was significantly (P ⁇ 0.05) blocked by the addition of excess unlabeled (NOTA-NCS)- HER3P1, indicating specific peptide binding, as shown in Figure 3C.
  • Parent peptide sequence C-L-P-T-K-F-R-S-C (SEQ ID NO.: 1) was mutated at positions 2-8 to an alanine, and binding of the phage to HER3 was assessed.
  • the results, shown in Figure 6, illustrate that for positions 2-4 and 7-8, substitution to alanine diminished binding to HER3 by 50% or greater. In contrast, positions 5-6 were unaffected by the mutation. This data demonstrates that amino acids 2-4 and 7-8 are involved in peptide binding, whereas 5 and 6 are amenable to mutation.
  • mice bearing human MDA-MB-453 tumors were injected with 68 Ga-(NOTA-NCS)-HER3Pl and subjected to PET imaging.
  • mice were treated with 100 mg/kg of lapatinib every 12 hours for 48 hours following PET imaging. At the end of 48 hours, mice were imaged in the exact same manner as before treatment.
  • SUVmean levels of HER3P1 increased to 1.35 ⁇ 0.59, as shown in Figure 7. Representative images from one of the mice is shown in Figure 8.
  • HER3P1 PET imaging will allow for non- invasive quantification of HER3 receptor expression level changes in response to PI3K inhibition, facilitating rapid analysis of HER3 mediated resistance across a panel of triple negative breast cancers (TNBCs).
  • TNBCs triple negative breast cancers
  • a panel of TNBC cell lines considered sensitive or resistant to PI3K inhibition with GDC-0941 (IC50 ⁇ 1 ⁇ ) (Sensitive: HDQ-P1, CAL-51, HCC-70, CAL-148, HCC- 1395, HS-578T, CAL-148; Resistant: MDA-MB-468, MDA-MB-231, MDA-MB-157, BT-549, CAL-120) (see e.g., O'Brien et al, Clin Cancer Res. 2010, 16:3670-3683) will be treated with GDC-0941.
  • the selected cell lines are reflective of the genetic heterogeneity of TNBC, with all dominant genetic categories represented (PI3KCA mutations, PTEN loss, KRAS mutant, EGFR over expression).
  • the cell lines will be treated with the PI3K inhibitor GDC-0941 and Western blot will be used to assess expression and phosphorylation status of HER3, as well as downstream growth signaling proteins (PI3K, AKT, mTOR, S6K, ERK) to assess for persistent signaling after PI3K inhibition.
  • whole cell viability and proliferation will also be analyzed using Celltiter-Glo. Eight cell lines will be selected for further in vivo analysis.
  • Cell lines will include two sensitive cell line without HER3 upregulation, two sensitive cell line demonstrating HER3 upregulation, two resistant cell lines without HER3 upregulation after treatment, and two resistant cell lines with HER3 upregulation after treatment.
  • xenografts of the selected tumors will be implanted in nu/nu female mice. Once tumors measure 5 mm in diameter, mice will be imaged by PET/CT with 68 GA-HER3P1 (i.e., 68 Ga-(NOTA-NCS)HER3Pl), according to our previously established protocols (see e.g., Wehrenberg-Klee et al, Journal of Nuclear Medicine, 2016, 57(9): 1413-1419).
  • mice will be randomized to 48 h treatment with 75 mg/kg GDC-0941 or vehicle. 48 h following treatment initiation, mice will be injected with 68 Ga-HER3Pl, and PET imaged again as described above. To confirm accurate quantification of HER3 expression by our HER3 PET probe, as well as similar biochemical response to PI3K inhibition in vivo as defined during in vitro cell analysis, mice will be sacrificed following final imaging and tumors harvested. Radioactivity of excised tumors will be immediately measured by gamma counter. Following radioactive decay, tumor will be homogenized, lysed and the tumor lysate analyzed by Western blot as above.
  • Example 9 Using Imaged HER3 Upregulation Patterns, Test Efficacy of HER3 Inhibitor Addition to Inhibit TNBC Growth in vitro and in vivo.
  • HER3 PET imaging From the eight cell lines identified for HER3 PET imaging in Example 8, a subset of four cell lines including one sensitive cell line without HER3 upregulation, one sensitive cell line demonstrating HER3 upregulation, one resistant cell line without HER3 upregulation after treatment, and one resistant cell line with HER3 upregulation after treatment will be analyzed in order to correlate imaging findings to proliferation and growth responses.
  • Each cell line will be treated with PI3K inhibitor, PI3K inhibitor with additional HER3 inhibitor, or vehicle. Efficacy of treatment regimens overtime will be assessed with viability (MTT, Presto Blue, WST-1) and clonogenic (Crystal violet) assays. Growth pathway inhibition will be assessed by Western blot.
  • the same cell lines selected for in vitro analysis will also be analyzed in vivo.
  • mice bearing cell line xenografts will be treated with vehicle, single agent PI3K inhibitor, or PI3K inhibitor with the addition of HER3 inhibitor. Tumor volumes will be measured three times a week. Experiments will terminate when significant differences in tumor growth are recorded between the treatment arms and/or the tumor volumes reach size requiring euthanasia. These experiments will also inform on treatment tolerability. Tumors will be harvested at the end of the experiments to confirm target inhibition. Conventionally, we will always collect tissue samples two hours from the last drug administration to allow both intra- and inter-experimental comparisons. When possible (xenografts are not too small and/or fibrotic), samples will be analyzed by Western blot against the total and phosphorylated forms of the targeted RTKs and signaling pathway effectors
  • FFPE paraffin- embedded
  • mice bearing PDXs Female nude mice bearing PDXs will be used to implant subsequent mice for all studies. Tumors will be imaged with 68 Ga-HER3Pl and randomized to treatment with GDC-0941 (PI3K inhibitor) or vehicle. Following two days of treatment, the mice will undergo repeat 68 Ga-HER3Pl imaging, as previously described, and tumoral HER3 PET SUV will be measured. The GDC-0941 group will then be randomized to receive additional HER3 inhibitor or additional vehicle. Following the initiation of secondary treatment, tumor volumes will be measured by caliper at least three times a week. Experiments will be carried out for 35 days following the beginning of secondary treatment, or when the tumor volumes reach unethical size. Tumors will be harvested at the end of the experiments to confirm target inhibition.
  • GDC-0941 PI3K inhibitor
  • samples will be analyzed ex vivo by Western blot when possible against the total and phosphorylated forms of HER3 as well as downstream signaling proteins as described above. IHC samples will also be analyzed following previously described technique.
  • a panel of TNBC cell lines considered sensitive or resistant to AKT inhibition with GDC-0068 (ICso ⁇ 1 ⁇ ) (Sensitive: CAL-148, HCC-70, CAL-51, HCC-1395, HS- 578T; Resistant: MDA-MB-468, HC- 143, CAL-85-1, HDQ-P1, MDA-MB-231, BT- 549)4,37 will be treated with GDC-0068.
  • the identical cell signaling and viability assays performed in Examples 8-10, will be performed in Example 11 to compare the effects of AKT inhibition. For each therapy, four cell lines will be selected for further in vivo analysis.
  • Cell lines will include a sensitive cell line without HER3 upregulation, a sensitive cell line demonstrating the most HER3 upregulation, a resistant cell line without HER3 upregulation after treatment, and a resistant cell line with HER3 upregulation after treatment.
  • xenografts of the selected tumors will be implanted in nu/nu female mice and PET images will be acquired and data analyzed in the same manner as detailed in Examples 8-10.
  • mice will be randomized to 48 h treatment with 75 mg/kg GDC-0068 or vehicle. 48 h following treatment initiation, mice will be injected with 68 Ga-HER3Pl and again imaged with image analysis as per Example 8.
  • mice will be sacrificed, tumors harvested and the tumor lysate analyzed by Western blot as above.
  • Example 12 Imaged HER3 Upregulation Patterns in Response to AKT Inhibition, Test Efficacy of HER3 Inhibitor Addition to Inhibit TNBC Growth Both in vitro and in vivo
  • Example 11 From the eight cell lines identified for HER3 PET imaging in Example 11 , a subset of four cell lines including one sensitive cell line without HER3 upregulation, one sensitive cell line demonstrating HER3 upregulation, one resistant cell line without HER3 upregulation after treatment, and one resistant cell line with HER3 upregulation after treatment will be analyzed in order to correlate imaging findings to proliferation and growth responses.
  • In vitro studies of treatment efficacy for HER3 inhibitor combination therapies will be systematically conducted as in Example 9. The same cell lines selected for in vitro analysis will also be analyzed in vivo. Female nu/nu mice bearing cell line xenografts will be treated with vehicle, single agent AKT inhibitor, or AKT inhibitor with the addition of HER3 inhibitor. Tumor volumes will be measured and following completion of study excised tumors will be assessed as in Example 9.
  • Example 13. TNBC PDX Tumor HER3 PET Imaging and Adaptive Therapy Following AKT Inhibition
  • TNBC PDX lines will be utilized, as outlined in Example 10. Tumors will be imaged with HER3P1 and then randomized to treatment with the AKT inhibitor GDC- 0068 75 mg/kg or vehicle. Following two days of treatment, the mice will undergo repeat imaging, as previously described, and tumoral HER3 PET SUV will be measured. GDC- 0068 treated tumors will then be randomized to receive HER3 inhibitor therapy or vehicle. Following the initiation of secondary treatment, tumor volumes will be measured for 35 days following the beginning of secondary treatment, or when the tumor volumes reach unethical size. Tumors will be harvested at the end of the experiments to confirm target inhibition.
  • AKT inhibitor GDC- 0068 75 mg/kg or vehicle Following two days of treatment, the mice will undergo repeat imaging, as previously described, and tumoral HER3 PET SUV will be measured. GDC- 0068 treated tumors will then be randomized to receive HER3 inhibitor therapy or vehicle. Following the initiation of secondary treatment, tumor volumes will be measured for 35 days following the beginning of secondary treatment, or when the tumor
  • samples will be analyzed ex vivo by Western blot when possible against the total and phosphorylated forms of HER3 as well as downstream signaling proteins as described above. IHC samples will also be analyzed following previously described technique.
  • HER2+ breast cancer cell lines either sensitive to HER2 inhibition (BT-474, SKBR3, UACC812, UACC893, ZR-75-30)47 or resistant to HER2 inhibition (JTMT-1, MDA-MB-453, HCC1569, HCC1954, MDA-MB-361) (see e.g., Wang et al, Breast Cancer Res. 2011, 13:R121), and including the dominant mutations found in HER2+ breast cancer (PI3KCA mutations, PTEN loss) will be treated with standard treatment dose of the HER2 inhibitor lapatinib (1 ⁇ ) or vehicle for 48 h.
  • HER3 and HER2 will be assessed by Western blot, and quantified as described above.
  • Cell viability (Celltiter-Glo) assays will be performed to monitor the effects of lapatinib treatment on proliferation. After in vitro analysis, a total of four cell lines will be selected for further in vivo imaging analysis. The identical cell signaling and viability assays performed in Examples 8-10, will be performed in this Example to compare the effects of AKT inhibition. For each therapy, four cell lines will be selected for further in vivo analysis.
  • Cell lines will include a sensitive cell line without HER3 upregulation, a sensitive cell line demonstrating the most HER3 upregulation, a resistant cell line without HER3 upregulation after treatment, and a resistant cell line with HER3 upregulation after treatment.
  • xenografts of the selected tumors will be implanted in nu/nu female mice, randomized to 48 h treatment with lapatinib or vehicle. Following treatment initiation, mice will be injected with dose of HER3 PET Probe. PET imaging data will be obtained and analyzed as described in Examples 8-10. Tumors will be excised and tumor and analyzed by Western blot for expression and phosphorylation of HER3 and downstream signaling proteins as described for in vitro studies.
  • Example 14 The four cell lines identified for HER3 PET imaging in Example 14 will be used for treatment experiments. For each cell line, in vitro studies of treatment efficacy for HER3 inhibitor and HER2 inhibitor combination regimens will be conducted in the same manner as Examples 9 and 12. Following in vitro analysis, groups of female nu/nu mice bearing cell-line xenografts will be treated with control, lapatinib, or the combination of lapatinib and HER3 inhibitor, tumor volumes measured, and when possible tumors excised an analyzed ex vivo to determine the molecular signatures of effective therapy and resistance outlined in Example 14.
  • HER2+ PDX lines will be established in a manner similar to Examples 10 and 13.
  • Female nude mice bearing PDXs will be PET imaged and then randomized to treatment with lapatinib or vehicle. Following two days of treatment, the mice will be undergo repeat HER3 PET imaging. The lapatinib arm will then be further randomized to receive either HER3 inhibitor therapy in addition to previously assigned therapy, or additional vehicle.
  • tumor volumes will be measured by caliper three times a week. Experiments will be carried out for 35 days following the beginning of secondary treatment, or when the tumor volumes reach unethical size. Tumors will be harvested at experiment end to confirm target inhibition. Samples will be analyzed ex vivo by Western blot when possible against the total and phosphorylated forms of HER3 as well as downstream signaling proteins as described above. IHC samples will also be analyzed following previously described technique.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Epidemiology (AREA)
  • Optics & Photonics (AREA)
  • Physics & Mathematics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
  • Organic Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

L'invention concerne des compositions utiles pour l'imagerie de HER3. Une composition donnée à titre d'exemple est utile en tant que radiotraceur pour une imagerie par tomographie par émission de positrons (TEP). L'invention concerne également des procédés d'imagerie de HER3 et des polythérapies comprenant les agents d'imagerie HER3.
PCT/US2017/069031 2016-12-29 2017-12-29 Peptides her3 pour imagerie et radiothérapie WO2018126183A2 (fr)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US16/475,020 US11241511B2 (en) 2016-12-29 2017-12-29 HER3 peptides for imaging and radiotherapy
US17/562,861 US20220175975A1 (en) 2016-12-29 2021-12-27 HER3 Peptides for Imaging and Radiotherapy

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201662440052P 2016-12-29 2016-12-29
US62/440,052 2016-12-29

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US16/475,020 A-371-Of-International US11241511B2 (en) 2016-12-29 2017-12-29 HER3 peptides for imaging and radiotherapy
US17/562,861 Division US20220175975A1 (en) 2016-12-29 2021-12-27 HER3 Peptides for Imaging and Radiotherapy

Publications (2)

Publication Number Publication Date
WO2018126183A2 true WO2018126183A2 (fr) 2018-07-05
WO2018126183A3 WO2018126183A3 (fr) 2018-08-23

Family

ID=62710947

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2017/069031 WO2018126183A2 (fr) 2016-12-29 2017-12-29 Peptides her3 pour imagerie et radiothérapie

Country Status (2)

Country Link
US (2) US11241511B2 (fr)
WO (1) WO2018126183A2 (fr)

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4957939A (en) 1981-07-24 1990-09-18 Schering Aktiengesellschaft Sterile pharmaceutical compositions of gadolinium chelates useful enhancing NMR imaging
US4472509A (en) 1982-06-07 1984-09-18 Gansow Otto A Metal chelate conjugated monoclonal antibodies
US4938948A (en) 1985-10-07 1990-07-03 Cetus Corporation Method for imaging breast tumors using labeled monoclonal anti-human breast cancer antibodies
US5329028A (en) 1992-08-05 1994-07-12 Genentech, Inc. Carbohydrate-directed cross-linking reagents
EP2100900A1 (fr) 2008-03-07 2009-09-16 Universitätsspital Basel Conjugués d'antagoniste de peptide analogue de bombésine
WO2011153050A1 (fr) 2010-06-02 2011-12-08 The Trustees Of The University Of Pennsylvania Méthodes et utilisation de composés qui se lient au complexe du récepteur her2/neu
CN104768976B (zh) 2012-10-05 2020-09-22 阿菲博迪公司 Her3结合多肽

Also Published As

Publication number Publication date
WO2018126183A3 (fr) 2018-08-23
US11241511B2 (en) 2022-02-08
US20200121814A1 (en) 2020-04-23
US20220175975A1 (en) 2022-06-09

Similar Documents

Publication Publication Date Title
KR102662725B1 (ko) Pd-l1 발현에 기반한 종양 및 면역 세포 영상화
CN109053616B (zh) 前列腺特异性膜抗原(psma)的标记的抑制剂及其用途
US11638765B2 (en) Double-labeled probe for molecular imaging and use thereof
EP3502122B1 (fr) Peptide cible de tumeur maligne
KR102665275B1 (ko) 신규 항인간 CEACAM5 항체 Fab 프래그먼트
US11452786B2 (en) Double-labeled probe for molecular imaging and use thereof
AU2018211539B2 (en) Compositions and methods for cancer imaging and radiotherapy
EP2904013B1 (fr) Polypeptides de liaison de her3
US20220175975A1 (en) HER3 Peptides for Imaging and Radiotherapy
EP2380597A1 (fr) Dérivés de cyclopeptide et ses utilisations
WO2022211051A1 (fr) Complexe radioactif d'anticorps anti-egfr, et produit radiopharmaceutique
JP2022549258A (ja) セラノスティックスとして使用するための放射性標識grprアンタゴニスト
KR20230174238A (ko) 방사성 항종양제
KR20220104703A (ko) 영상화 및 치료용 피브린-결합 화합물
Dobitz Oligoprolines as Multivalent Scaffolds for Tumor Targeting
EA042953B1 (ru) Композиции и способы для визуализации и лучевой терапии рака
Do Integrin targeted imaging agents

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 17889322

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 17889322

Country of ref document: EP

Kind code of ref document: A2